Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.736
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(25): e2404457121, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38865275

RESUMEN

The fat mass and obesity-associated fatso (FTO) protein is a member of the Alkb family of dioxygenases and catalyzes oxidative demethylation of N6-methyladenosine (m6A), N1-methyladenosine (m1A), 3-methylthymine (m3T), and 3-methyluracil (m3U) in single-stranded nucleic acids. It is well established that the catalytic activity of FTO proceeds via two coupled reactions. The first reaction involves decarboxylation of alpha-ketoglutarate (αKG) and formation of an oxyferryl species. In the second reaction, the oxyferryl intermediate oxidizes the methylated nucleic acid to reestablish Fe(II) and the canonical base. However, it remains unclear how binding of the nucleic acid activates the αKG decarboxylation reaction and why FTO demethylates different methyl modifications at different rates. Here, we investigate the interaction of FTO with 5-mer DNA oligos incorporating the m6A, m1A, or m3T modifications using solution NMR, molecular dynamics (MD) simulations, and enzymatic assays. We show that binding of the nucleic acid to FTO activates a two-state conformational equilibrium in the αKG cosubstrate that modulates the O2 accessibility of the Fe(II) catalyst. Notably, the substrates that provide better stabilization to the αKG conformation in which Fe(II) is exposed to O2 are demethylated more efficiently by FTO. These results indicate that i) binding of the methylated nucleic acid is required to expose the catalytic metal to O2 and activate the αKG decarboxylation reaction, and ii) the measured turnover of the demethylation reaction (which is an ensemble average over the entire sample) depends on the ability of the methylated base to favor the Fe(II) state accessible to O2.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Hierro , Ácidos Cetoglutáricos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/química , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/química , Hierro/metabolismo , Hierro/química , Humanos , Especificidad por Sustrato , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/química , Conformación Proteica , Uracilo/metabolismo , Uracilo/análogos & derivados , Uracilo/química , Simulación de Dinámica Molecular , Timina/análogos & derivados
2.
Sci Rep ; 14(1): 11609, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773214

RESUMEN

No biomarker has yet been identified that allows accurate diagnosis and prognosis of oral cancers. In this study, we investigated the presence of key metabolites in oral cancer using proton nuclear magnetic resonance (NMR) spectroscopy to identify metabolic biomarkers of gingivobuccal oral squamous cell carcinoma (GB-OSCC). NMR spectroscopy revealed that uracil was expressed in 83.09% of tumor tissues and pyrimidine metabolism was active in GB-OSCC; these results correlated well with immunohistochemistry (IHC) and RNA sequencing data. Based on further gene and protein analyses, we proposed a pathway for the production of uracil in GB-OSCC tissues. Uridinetriphosphate (UTP) is hydrolyzed to uridine diphosphate (UDP) by CD39 in the tumor microenvironment (TME). We hypothesized that UDP enters the cell with the help of the UDP-specific P2Y6 receptor for further processing by ENTPD4/5 to produce uracil. As the ATP reserves diminish, the weakened immune cells in the TME utilize pyrimidine metabolism as fuel for antitumor activity, and the same mechanism is hijacked by the tumor cells to promote their survival. Correspondingly, the differential expression of ENTPD4 and ENTPD5 in immune and tumor cells, respectively, indicatedtheir involvement in disease progression. Furthermore, higher uracil levels were detected in patients with lymph node metastasis, indicating that metastatic potential is increased in the presence of uracil. The presence of uracil and/or expression patterns of intermediate molecules in purine and pyrimidine pathways, such asCD39, CD73, and P2Y6 receptors together with ENTPD4 and ENTPD5, hold promise as biomarker(s) for oral cancer diagnosis and prognosis.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Boca , Pirimidinas , Uracilo , Humanos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Uracilo/metabolismo , Biomarcadores de Tumor/metabolismo , Pirimidinas/metabolismo , Femenino , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Masculino , Persona de Mediana Edad , Microambiente Tumoral , Anciano , Apirasa/metabolismo
3.
J Chem Theory Comput ; 20(10): 4218-4228, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38720241

RESUMEN

iso-Orotate decarboxylase (IDCase), which is involved in the thymidine salvage pathway, has attracted considerable interest owing to its chemical similarity to a hypothetical DNA decarboxylase in mammals. Although valuable insights into the active DNA demethylation of 5-methyl-cytosine can be obtained from the decarboxylation mechanism of 5-carboxyl-uracil (5caU) catalyzed by IDCase, this mechanism remains under debate. In this study, the catalytic mechanism of 5caU decarboxylation by IDCase was studied using hybrid quantum mechanics/molecular mechanics (QM/MM) methodologies and density functional theory (DFT) calculations with a truncated model. The calculations supported a mechanism involving three sequential stages: activation of the 5caU substrate via proton transfer from an arginine (R262') to the carboxyl group of 5caU, formation of a tetrahedral intermediate, and decarboxylation of the tetrahedral intermediate to generate uracil as the product. The reaction pathways and structures obtained using the QM/MM and DFT methods coincided with each other. These simulations provided detailed insights into the unique mechanism of IDCase, clarifying various unresolved issues, such as the critical role of R262'. In addition, aspartate D323 was found to act as a general base in the tetrahedral intermediate formation step and a general acid in the later C-C bond cleavage step.


Asunto(s)
Teoría Funcional de la Densidad , Descarboxilación , Simulación de Dinámica Molecular , Teoría Cuántica , Carboxiliasas/química , Carboxiliasas/metabolismo , Biocatálisis , Orotidina-5'-Fosfato Descarboxilasa/química , Orotidina-5'-Fosfato Descarboxilasa/metabolismo , Uracilo/química , Uracilo/metabolismo
4.
Chem Biol Interact ; 397: 111046, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38735451

RESUMEN

Cylindrospermopsin, a potent hepatotoxin produced by harmful cyanobacterial blooms, poses environmental and human health concerns. We used a 3D human liver in vitro model based on spheroids of HepG2 cells, in combination with molecular and biochemical assays, automated imaging, targeted LC-MS-based proteomics, and lipidomics, to explore cylindrospermopsin effects on lipid metabolism and the processes implicated in hepatic steatosis. Cylindrospermopsin (1 µM, 48 h) did not significantly affect cell viability but partially reduced albumin secretion. However, it increased neutral lipid accumulation in HepG2 spheroids while decreasing phospholipid levels. Simultaneously, cylindrospermopsin upregulated genes for lipogenesis regulation (SREBF1) and triacylglycerol synthesis (DGAT1/2) and downregulated genes for fatty acid synthesis (ACLY, ACCA, FASN, SCD1). Fatty acid uptake, oxidation, and lipid efflux genes were not significantly affected. Targeted proteomics revealed increased levels of perilipin 2 (adipophilin), a major hepatocyte lipid droplet-associated protein. Lipid profiling quantified 246 lipid species in the spheroids, with 28 significantly enriched and 15 downregulated by cylindrospermopsin. Upregulated species included neutral lipids, sphingolipids (e.g., ceramides and dihexosylceramides), and some glycerophospholipids (phosphatidylethanolamines, phosphatidylserines), while phosphatidylcholines and phosphatidylinositols were mostly reduced. It suggests that cylindrospermopsin exposures might contribute to developing and progressing towards hepatic steatosis or metabolic dysfunction-associated steatotic liver disease (MASLD).


Asunto(s)
Alcaloides , Toxinas Bacterianas , Toxinas de Cianobacterias , Metabolismo de los Lípidos , Hígado , Uracilo , Humanos , Alcaloides/farmacología , Toxinas Bacterianas/metabolismo , Uracilo/análogos & derivados , Uracilo/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Células Hep G2 , Hígado/metabolismo , Hígado/efectos de los fármacos , Homeostasis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Proteómica , Lipidómica , Lipogénesis/efectos de los fármacos
5.
Commun Biol ; 7(1): 529, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704509

RESUMEN

Intra-organism biodiversity is thought to arise from epigenetic modification of constituent genes and post-translational modifications of translated proteins. Here, we show that post-transcriptional modifications, like RNA editing, may also contribute. RNA editing enzymes APOBEC3A and APOBEC3G catalyze the deamination of cytosine to uracil. RNAsee (RNA site editing evaluation) is a computational tool developed to predict the cytosines edited by these enzymes. We find that 4.5% of non-synonymous DNA single nucleotide polymorphisms that result in cytosine to uracil changes in RNA are probable sites for APOBEC3A/G RNA editing; the variant proteins created by such polymorphisms may also result from transient RNA editing. These polymorphisms are associated with over 20% of Medical Subject Headings across ten categories of disease, including nutritional and metabolic, neoplastic, cardiovascular, and nervous system diseases. Because RNA editing is transient and not organism-wide, future work is necessary to confirm the extent and effects of such editing in humans.


Asunto(s)
Desaminasas APOBEC , Citidina Desaminasa , Edición de ARN , Humanos , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , Polimorfismo de Nucleótido Simple , Citosina/metabolismo , Desaminasa APOBEC-3G/metabolismo , Desaminasa APOBEC-3G/genética , Uracilo/metabolismo , Proteínas/genética , Proteínas/metabolismo , Citosina Desaminasa/genética , Citosina Desaminasa/metabolismo
6.
Sci Total Environ ; 929: 172590, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38642746

RESUMEN

Harmful cyanobacterial blooms have increased globally, releasing hazardous cyanotoxins that threaten the safety of water resources. Constructed wetlands (CWs) are a nature-based and low-cost solution to purify and remove cyanotoxins from water. However, bio-mechanistic understanding of the biotransformation processes expected to drive cyanotoxin removal in such systems is poor, and primarily focused on bacteria. Thus, the present study aimed at exploring the fungal contribution to microcystin-LR and cylindrospermopsin biodegradation in CWs. Based on CW mesocosms, two experimental approaches were taken: a) amplicon sequencing studies were conducted to investigate the involvement of the fungal community; and b) CW fungal isolates were tested for their microcystin-LR and cylindrospermopsin degradation capabilities. The data uncovered effects of seasonality (spring or summer), cyanotoxin exposure, vegetation (unplanted, Juncus effusus or Phragmites australis) and substratum (sand or gravel) on the fungal community structure. Additionally, the arbuscular mycorrhizal fungus Rhizophagus and the endophyte Myrmecridium showed positive correlations with cyanotoxin removal. Fungal isolates revealed microcystin-LR-removal potentials of approximately 25 % in in vitro biodegradation experiments, while the extracellular chemical fingerprint of the cultures suggested a potential intracellular metabolization. The results from this study may help us understand the fungal contribution to cyanotoxin removal, as well as their ecology in CWs.


Asunto(s)
Biodegradación Ambiental , Hongos , Microcistinas , Humedales , Microcistinas/metabolismo , Hongos/metabolismo , Toxinas Bacterianas/metabolismo , Alcaloides/metabolismo , Toxinas de Cianobacterias , Toxinas Marinas/metabolismo , Contaminantes Químicos del Agua/metabolismo , Eliminación de Residuos Líquidos/métodos , Uracilo/análogos & derivados , Uracilo/metabolismo
7.
Mol Cell ; 84(11): 2036-2052.e7, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38688279

RESUMEN

Alterations of bases in DNA constitute a major source of genomic instability. It is believed that base alterations trigger base excision repair (BER), generating DNA repair intermediates interfering with DNA replication. Here, we show that genomic uracil, a common type of base alteration, induces DNA replication stress (RS) without being processed by BER. In the absence of uracil DNA glycosylase (UNG), genomic uracil accumulates to high levels, DNA replication forks slow down, and PrimPol-mediated repriming is enhanced, generating single-stranded gaps in nascent DNA. ATR inhibition in UNG-deficient cells blocks the repair of uracil-induced gaps, increasing replication fork collapse and cell death. Notably, a subset of cancer cells upregulates UNG2 to suppress genomic uracil and limit RS, and these cancer cells are hypersensitive to co-treatment with ATR inhibitors and drugs increasing genomic uracil. These results reveal unprocessed genomic uracil as an unexpected source of RS and a targetable vulnerability of cancer cells.


Asunto(s)
Reparación del ADN , Replicación del ADN , Inestabilidad Genómica , Uracil-ADN Glicosidasa , Uracilo , Humanos , Uracilo/metabolismo , Uracil-ADN Glicosidasa/metabolismo , Uracil-ADN Glicosidasa/genética , Reparación del ADN/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Daño del ADN , Línea Celular Tumoral , Neoplasias/genética , Neoplasias/patología , Neoplasias/metabolismo
8.
J Microbiol Biotechnol ; 34(5): 1178-1187, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38563100

RESUMEN

Cordyceps militaris is a significant edible fungus that produces a variety of bioactive compounds. We have previously established a uridine/uracil auxotrophic mutant and a corresponding Agrobacterium tumefaciens-mediated transformation (ATMT) system for genetic characterization in C. militaris using pyrG as a screening marker. In this study, we constructed an ATMT system based on a dual pyrG and hisB auxotrophic mutant of C. militaris. Using the uridine/uracil auxotrophic mutant as the background and pyrG as a selection marker, the hisB gene encoding imidazole glycerophosphate dehydratase, required for histidine biosynthesis, was knocked out by homologous recombination to construct a histidine auxotrophic C. militaris mutant. Then, pyrG in the histidine auxotrophic mutant was deleted to construct a ΔpyrG ΔhisB dual auxotrophic mutant. Further, we established an ATMT transformation system based on the dual auxotrophic C. militaris by using GFP and DsRed as reporter genes. Finally, to demonstrate the application of this dual transformation system for studies of gene function, knock out and complementation of the photoreceptor gene CmWC-1 in the dual auxotrophic C. militaris were performed. The newly constructed ATMT system with histidine and uridine/uracil auxotrophic markers provides a promising tool for genetic modifications in the medicinal fungus C. militaris.


Asunto(s)
Agrobacterium tumefaciens , Cordyceps , Transformación Genética , Uracilo , Agrobacterium tumefaciens/genética , Agrobacterium tumefaciens/metabolismo , Cordyceps/genética , Cordyceps/metabolismo , Cordyceps/crecimiento & desarrollo , Uracilo/metabolismo , Histidina/metabolismo , Uridina/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Técnicas de Inactivación de Genes , Hidroliasas/genética , Hidroliasas/metabolismo , Genes Reporteros , Mutación , Recombinación Homóloga
9.
RNA ; 30(7): 901-919, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38670632

RESUMEN

A key to understanding the roles of RNA in regulating gene expression is knowing their structures in vivo. One way to obtain this information is through probing the structures of RNA with chemicals. To probe RNA structure directly in cells, membrane-permeable reagents that modify the Watson-Crick (WC) face of unpaired nucleotides can be used. Although dimethyl sulfate (DMS) has led to substantial insight into RNA structure, it has limited nucleotide specificity in vivo, with WC face reactivity only at adenine (A) and cytosine (C) at neutral pH. The reagent 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) was recently shown to modify the WC face of guanine (G) and uracil (U). Although useful at lower concentrations in experiments that measure chemical modifications by reverse transcription (RT) stops, at higher concentrations necessary for detection by mutational profiling (MaP), EDC treatment leads to degradation of RNA. Here, we demonstrate EDC-stimulated degradation of RNA in Gram-negative and Gram-positive bacteria. In an attempt to overcome these limitations, we developed a new carbodiimide reagent, 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide methiodide (ETC), which we show specifically modifies unpaired Gs and Us in vivo without substantial degradation of RNA. We establish ETC as a probe for MaP and optimize the RT conditions and computational analysis in Escherichia coli Importantly, we demonstrate the utility of ETC as a probe for improving RNA structure prediction both alone and with DMS.


Asunto(s)
Guanina , Conformación de Ácido Nucleico , Ésteres del Ácido Sulfúrico , Uracilo , Ésteres del Ácido Sulfúrico/química , Uracilo/química , Uracilo/análogos & derivados , Uracilo/metabolismo , Guanina/química , Guanina/metabolismo , ARN/química , ARN/genética , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Carbodiimidas/química , ARN Bacteriano/química , ARN Bacteriano/genética , Estabilidad del ARN , Indicadores y Reactivos/química
10.
BMC Biotechnol ; 24(1): 17, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566117

RESUMEN

Thermostable DNA polymerases, such as Taq isolated from the thermophilic bacterium Thermus aquaticus, enable one-pot exponential DNA amplification known as polymerase chain reaction (PCR). However, properties other than thermostability - such as fidelity, processivity, and compatibility with modified nucleotides - are important in contemporary molecular biology applications. Here, we describe the engineering and characterization of a fusion between a DNA polymerase identified in the marine archaea Nanoarchaeum equitans and a DNA binding domain from the thermophile Sulfolobus solfataricus. The fusion creates a highly active enzyme, Neq2X7, capable of amplifying long and GC-rich DNA, unaffected by replacing dTTP with dUTP in PCR, and tolerant to various known PCR inhibitors. This makes it an attractive DNA polymerase for use, e.g., with uracil excision (USER) DNA assembly and for contamination-free diagnostics. Using a magnification via nucleotide imbalance fidelity assay, Neq2X7 was estimated to have an error rate lower than 2 ∙ 10-5 bp-1 and an approximately 100x lower fidelity than the parental variant Neq2X, indicating a trade-off between fidelity and processivity - an observation that may be of importance for similarly engineered DNA polymerases. Neq2X7 is easy to produce for routine application in any molecular biology laboratory, and the expression plasmid is made freely available.


Asunto(s)
ADN Polimerasa Dirigida por ADN , Uracilo , Reacción en Cadena de la Polimerasa , ADN Polimerasa Dirigida por ADN/genética , Uracilo/metabolismo , Plásmidos , ADN
11.
J Biol Chem ; 300(5): 107229, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38537698

RESUMEN

Mucosal-associated invariant T (MAIT) cells can elicit immune responses against riboflavin-based antigens presented by the evolutionary conserved MHC class I related protein, MR1. While we have an understanding of the structural basis of human MAIT cell receptor (TCR) recognition of human MR1 presenting a variety of ligands, how the semi-invariant mouse MAIT TCR binds mouse MR1-ligand remains unknown. Here, we determine the crystal structures of 2 mouse TRAV1-TRBV13-2+ MAIT TCR-MR1-5-OP-RU ternary complexes, whose TCRs differ only in the composition of their CDR3ß loops. These mouse MAIT TCRs mediate high affinity interactions with mouse MR1-5-OP-RU and cross-recognize human MR1-5-OP-RU. Similarly, a human MAIT TCR could bind mouse MR1-5-OP-RU with high affinity. This cross-species recognition indicates the evolutionary conserved nature of this MAIT TCR-MR1 axis. Comparing crystal structures of the mouse versus human MAIT TCR-MR1-5-OP-RU complexes provides structural insight into the conserved nature of this MAIT TCR-MR1 interaction and conserved specificity for the microbial antigens, whereby key germline-encoded interactions required for MAIT activation are maintained. This is an important consideration for the development of MAIT cell-based therapeutics that will rely on preclinical mouse models of disease.


Asunto(s)
Antígenos de Histocompatibilidad Clase I , Antígenos de Histocompatibilidad Menor , Células T Invariantes Asociadas a Mucosa , Ribitol , Animales , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Antígenos de Histocompatibilidad Menor/química , Ratones , Células T Invariantes Asociadas a Mucosa/inmunología , Células T Invariantes Asociadas a Mucosa/metabolismo , Humanos , Ribitol/análogos & derivados , Ribitol/metabolismo , Ribitol/química , Uracilo/análogos & derivados , Uracilo/metabolismo , Uracilo/química , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Cristalografía por Rayos X
12.
J Biosci Bioeng ; 137(6): 413-419, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38485553

RESUMEN

Uracil-thymine dehydrogenase (UTDH), which catalyzes the irreversible oxidation of uracil to barbituric acid in oxidative pyrimidine metabolism, was purified from Rhodococcus erythropolis JCM 3132. The finding of unusual stabilizing conditions (pH 11, in the presence of NADP+ or NADPH) enabled the enzyme purification. The purified enzyme was a heteromer consisting of three different subunits. The enzyme catalyzed oxidation of uracil to barbituric acid with artificial electron acceptors such as methylene blue, phenazine methosulfate, benzoquinone, and α-naphthoquinone; however, NAD+, NADP+, flavin adenine dinucleotide, and flavin mononucleotide did not serve as electron acceptors. The enzyme acted not only on uracil and thymine but also on 5-halogen-substituted uracil and hydroxypyrimidine (pyrimidone), while dihydropyrimidine, which is an intermediate in reductive pyrimidine metabolism, and purine did not serve as substrates. The activity of UTDH was enhanced by cerium ions, and this activation was observed with all combinations of substrates and electron acceptors.


Asunto(s)
Oxidación-Reducción , Pirimidinas , Rhodococcus , Uracilo , Uracilo/metabolismo , Uracilo/química , Pirimidinas/metabolismo , Rhodococcus/enzimología , NADP/metabolismo , Azul de Metileno/metabolismo , Azul de Metileno/química , Barbitúricos/metabolismo , Barbitúricos/química , Benzoquinonas/metabolismo , Benzoquinonas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Concentración de Iones de Hidrógeno , Timina/metabolismo , Timina/química , Especificidad por Sustrato , Metosulfato de Metilfenazonio/metabolismo , Metosulfato de Metilfenazonio/química
13.
Nat Commun ; 15(1): 2369, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38499553

RESUMEN

The APOBEC3 enzymes convert cytosines in single-stranded DNA to uracils to protect against viruses and retrotransposons but can contribute to mutations that diversify tumors. To understand the mechanism of mutagenesis, we map the uracils resulting from expression of APOBEC3B or its catalytic carboxy-terminal domain (CTD) in Escherichia coli. Like APOBEC3A, the uracilomes of A3B and A3B-CTD show a preference to deaminate cytosines near transcription start sites and the lagging-strand replication templates and in hairpin loops. Both biochemical activities of the enzymes and genomic uracil distribution show that A3A prefers 3 nt loops the best, while A3B prefers 4 nt loops. Reanalysis of hairpin loop mutations in human tumors finds intrinsic characteristics of both the enzymes, with a much stronger contribution from A3A. We apply Hairpin Signatures 1 and 2, which define A3A and A3B preferences respectively and are orthogonal to published methods, to evaluate their contribution to human tumor mutations.


Asunto(s)
Citosina , Neoplasias , Humanos , Citosina/metabolismo , Proteínas/metabolismo , Mutación , Citidina Desaminasa/metabolismo , Neoplasias/genética , Uracilo/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo
14.
ACS Synth Biol ; 13(4): 1303-1311, 2024 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-38529630

RESUMEN

In this study, we proposed a biological approach to efficiently produce pseudouridine (Ψ) from glucose and uracil in vivo using engineered Escherichia coli. By screening host strains and core enzymes, E. coli MG1655 overexpressing Ψ monophosphate (ΨMP) glycosidase and ΨMP phosphatase was obtained, which displayed the highest Ψ concentration. Then, optimization of the RBS sequences, enhancement of ribose 5-phosphate supply in the cells, and overexpression of the membrane transport protein UraA were investigated. Finally, fed-batch fermentation of Ψ in a 5 L fermentor can reach 27.5 g/L with a yield of 89.2 mol % toward uracil and 25.6 mol % toward glucose within 48 h, both of which are the highest to date. In addition, the Ψ product with a high purity of 99.8% can be purified from the fermentation broth after crystallization. This work provides an efficient and environmentally friendly protocol for allowing for the possibility of Ψ bioproduction on an industrial scale.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Escherichia coli/metabolismo , Seudouridina/metabolismo , Glucosa/metabolismo , Uracilo/metabolismo , Reactores Biológicos , Fermentación , Ingeniería Metabólica , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Escherichia coli/metabolismo
15.
Nucleic Acids Res ; 52(2): 784-800, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38000394

RESUMEN

Activation-induced cytidine deaminase (AID) interacts with replication protein A (RPA), the major ssDNA-binding protein, to promote deamination of cytosine to uracil in transcribed immunoglobulin (Ig) genes. Uracil-DNA glycosylase (UNG) acts in concert with AID during Ig diversification. In addition, UNG preserves genome integrity by base-excision repair (BER) in the overall genome. How UNG is regulated to support both mutagenic processing and error-free repair remains unknown. UNG is expressed as two isoforms, UNG1 and UNG2, which both contain an RPA-binding helix that facilitates uracil excision from RPA-coated ssDNA. However, the impact of this interaction in antibody diversification and genome maintenance has not been investigated. Here, we generated B-cell clones with targeted mutations in the UNG RPA-binding motif, and analysed class switch recombination (CSR), mutation frequency (5' Ig Sµ), and genomic uracil in clones representing seven Ung genotypes. We show that the UNG:RPA interaction plays a crucial role in both CSR and repair of AID-induced uracil at the Ig loci. By contrast, the interaction had no significant impact on total genomic uracil levels. Thus, RPA coordinates UNG during CSR and pre-replicative repair of mutagenic uracil in ssDNA but is not essential in post-replicative and canonical BER of uracil in dsDNA.


Asunto(s)
Proteína de Replicación A , Uracil-ADN Glicosidasa , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Reparación del ADN/genética , ADN de Cadena Simple/genética , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Inmunoglobulinas/genética , Mutágenos , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Uracilo/metabolismo , Uracil-ADN Glicosidasa/genética , Uracil-ADN Glicosidasa/metabolismo , Humanos , Animales , Ratones
16.
Microb Cell Fact ; 22(1): 255, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38087285

RESUMEN

BACKGROUND: Uridyl peptide compounds are renowned as a subclass of nucleoside antibiotics for their highly specific antibacterial activity against Gram-negative bacteria and the unique target of action. We previously activated the biosynthetic gene cluster of a uridyl peptide antibiotic, mureidomycin, in Streptomyces roseosporus NRRL 15998 by introducing an exogenous positive regulator gene ssaA, and the generated strain was designated as Sr-hA. This study aims to further explore mureidomycin analogs from Sr-hA as well as the collaborative roles of two wide-spread genes, SSGG-02980 and SSGG-03002 encoding putative nuclease/phosphatase and oxidoreductase respectively, in mureidomycin diversification. RESULTS: In order to understand how SSGG-02980 and SSGG-03002 contribute to mureidomycin biosynthesis, the gene disruption mutants and complementary strains were constructed. Mass spectrometry analyses revealed that two series of pairwise mureidomycin analogs were synthesized in Sr-hA with a two-dalton difference in molecular weight for each pair. By disruption of SSGG-03002, only mureidomycins with lower molecular weight (MRDs, 1-6) could be specifically accumulated in the mutant (∆03002-hA), whereas the other series of products with molecular weight plus 2 Da (rMRDs, 1'-6') became dominant in SSGG-02980 disruption mutant (∆02980-hA). Further comprehensive NMR analyses were performed to elucidate the structures, and three MRDs (3, 4, 5) with unsaturated double bond at C5-C6 of uracil group were characterized from ∆03002-hA. In contrast, the paired rMRDs analogs (3', 4', 5') from ∆SSGG-02980 corresponding to 3, 4 and 5 were shown to contain a single bond at this position. The results verified that SSGG-03002 participates in the reduction of uracil ring, whereas SSGG-02980 antagonizes the effect of SSGG-03002, which has been rarely recognized for a phosphatase. CONCLUSIONS: Overall, this study revealed the key roles of two wide-spread families of enzymes in Streptomyces. Of them, oxidoreductase, SSGG-03002, is involved in dihydro-mureidomycin biosynthesis of S. roseosporus, whereas nuclease/phosphatase, SSGG-02980, has an adverse effect on SSGG-03002. This kind of unusual regulation model between nuclease/phosphatase and oxidoreductase is unprecedented, providing new insights into the biosynthesis of mureidomycins in Streptomyces. The findings would be of significance for structural diversification of more uridyl peptide antibiotics against Gram-negative bacteria.


Asunto(s)
Antibacterianos , Streptomyces , Péptidos/metabolismo , Proteínas Bacterianas/metabolismo , Streptomyces/metabolismo , Oxidorreductasas/metabolismo , Uracilo/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Familia de Multigenes
17.
Insect Biochem Mol Biol ; 163: 104029, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37907139

RESUMEN

Dextran sulfate sodium is used in inflammatory bowel disease (IBD) mice models to trigger chronic intestinal inflammation. In this study, we have analyzed DSS effects in the genetic model and pest beetle, Tribolium castaneum, which can be easily and cost-effectively cultivated and examined in very large quantities compensating for individual variations. We fed the larvae with DSS and uracil, which is known to induce the production of reactive oxygen species by activating DUOX, a member of the NADPH oxidase family. Both chemicals induced IBD-like phenotypes, including impaired growth and development, midgut thickening, epithelial swelling, and a loss of epithelial barrier function. RNAi mediated knockdown of DUOX expression enhanced the effects of DSS and uracil on mortality. Finally, we showed that both treatments result in an altered activity of the intestinal microbiome, similar as observed in IBD patients. Our findings suggest that both chemicals impair the epithelial barrier by increasing the permeability of the peritrophic matrix. The loss of the barrier function may facilitate the entry of midgut bacteria triggering innate immune responses that also affect the intestinal microbiome. As the observed effects resemble those induced by DSS treatment in mice, T. castaneum might be suitable high-throughput invertebrate model for IBD research and preclinical studies.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Tribolium , Ratones , Animales , Tribolium/metabolismo , Sulfato de Dextran/metabolismo , Sulfato de Dextran/farmacología , Quitina/metabolismo , Uracilo/metabolismo , Uracilo/farmacología , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo
18.
J Mol Biol ; 435(19): 168226, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37544358

RESUMEN

Transporters mediate the uptake of solutes, metabolites and drugs across the cell membrane. The eukaryotic FurE nucleobase/H+ symporter of Aspergillus nidulans has been used as a model protein to address structure-function relationships in the APC transporter superfamily, members of which are characterized by the LeuT-fold and seem to operate by the so-called 'rocking-bundle' mechanism. In this study, we reveal the binding mode, translocation and release pathway of uracil/H+ by FurE using path collective variable, funnel metadynamics and rational mutational analysis. Our study reveals a stepwise, induced-fit, mechanism of ordered sequential transport of proton and uracil, which in turn suggests that FurE, functions as a multi-step gated pore, rather than employing 'rocking' of compact domains, as often proposed for APC transporters. Finally, our work supports that specific residues of the cytoplasmic N-tail are involved in substrate translocation, in line with their essentiality for FurE function.


Asunto(s)
Proteínas de Transporte de Membrana , Uracilo , Transporte Biológico , Membrana Celular/metabolismo , Transporte Iónico , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Protones , Uracilo/metabolismo
19.
Cell Metab ; 35(8): 1356-1372.e5, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37473754

RESUMEN

Liver mitochondria undergo architectural remodeling that maintains energy homeostasis in response to feeding and fasting. However, the specific components and molecular mechanisms driving these changes and their impact on energy metabolism remain unclear. Through comparative mouse proteomics, we found that fasting induces strain-specific mitochondrial cristae formation in the liver by upregulating MIC19, a subunit of the MICOS complex. Enforced MIC19 expression in the liver promotes cristae formation, mitochondrial respiration, and fatty acid oxidation while suppressing gluconeogenesis. Mice overexpressing hepatic MIC19 show resistance to diet-induced obesity and improved glucose homeostasis. Interestingly, MIC19 overexpressing mice exhibit elevated energy expenditure and increased pedestrian locomotion. Metabolite profiling revealed that uracil accumulates in the livers of these mice due to increased uridine phosphorylase UPP2 activity. Furthermore, uracil-supplemented diet increases locomotion in wild-type mice. Thus, MIC19-induced mitochondrial cristae formation in the liver increases uracil as a signal to promote locomotion, with protective effects against diet-induced obesity.


Asunto(s)
Metabolismo Energético , Hígado , Caminata , Mitocondrias Hepáticas/metabolismo , Hígado/metabolismo , Proteínas Mitocondriales/metabolismo , Proteoma/metabolismo , Animales , Ratones , Dieta Alta en Grasa , Aumento de Peso , Uracilo/metabolismo
20.
Nat Struct Mol Biol ; 30(8): 1153-1159, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37460895

RESUMEN

The interbacterial deaminase toxin DddA catalyzes cytosine-to-uracil conversion in double-stranded (ds) DNA and enables CRISPR-free mitochondrial base editing, but the molecular mechanisms underlying its unique substrate selectivity have remained elusive. Here, we report crystal structures of DddA bound to a dsDNA substrate containing the 5'-TC target motif. These structures show that DddA binds to the minor groove of a sharply bent dsDNA and engages the target cytosine extruded from the double helix. DddA Phe1375 intercalates in dsDNA and displaces the 5' (-1) thymine, which in turn replaces the target (0) cytosine and forms a noncanonical T-G base pair with the juxtaposed guanine. This tandem displacement mechanism allows DddA to locate a target cytosine without flipping it into the active site. Biochemical experiments demonstrate that DNA base mismatches enhance the DddA deaminase activity and relax its sequence selectivity. On the basis of the structural information, we further identified DddA mutants that exhibit attenuated activity or altered substrate preference. Our studies may help design new tools useful in genome editing or other applications.


Asunto(s)
Citosina , ADN , Citosina/metabolismo , Desaminación , Secuencia de Bases , ADN/química , Uracilo/metabolismo , Citidina Desaminasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...